Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 271(Pt 1): 132518, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777025

ABSTRACT

Chondroitinases play important roles in structural and functional studies of chondroitin sulfates. Carbohydrate-binding module (CBM) is generally considered as an accessory module in carbohydrate-active enzymes, which promotes the association of the appended enzyme with the substrate and potentiates the catalytic activity. However, the role of natural CBM in chondroitinases has not been investigated. Herein, a novel chondroitinase ChABC29So containing an unknown domain with a predicted ß-sandwich fold was discovered from Segatella oris. Recombinant ChABC29So showed enzyme activity towards chondroitin sulfates and hyaluronic acid and acted in a random endo-acting manner. The unknown domain exhibited a chondroitin sulfate-binding capacity and was identified as a CBM. Biochemical characterization of ChABC29So and the CBM-truncated enzyme revealed that the CBM enhances the catalytic activity, thermostability, and disaccharide proportion in the final enzymatic products of ChABC29So. These findings demonstrate the role of the natural CBM in a chondroitinase and will guide future modification of chondroitinases.


Subject(s)
Chondroitin ABC Lyase , Chondroitin Sulfates , Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Chondroitin ABC Lyase/genetics , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Substrate Specificity , Enzyme Stability , Protein Binding , Amino Acid Sequence , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism
2.
Appl Environ Microbiol ; 88(22): e0154622, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36342199

ABSTRACT

The degradation of glycosaminoglycans (GAGs) by intestinal bacteria is critical for their colonization in the human gut and the health of the host. Both colonic Bacteroides and Firmicutes have been reported to degrade GAGs; however, the enzymatic details of the latter remain largely unknown. Our bioinformatic analyses of fecal Firmicutes revealed that their genomes, especially Hungatella hathewayi strains, are an abundant source of putative GAG-specific catabolic enzymes. Subsequently, we isolated a Firmicutes strain, H. hathewayi N2-326, that can catabolize various GAGs. While H. hathewayi N2-326 was as efficient in utilizing chondroitin sulfate A (CSA) and dermatan sulfate as Bacteroides thetaiotaomicron, a well-characterized GAG degrader, it outperformed B. thetaiotaomicron in assimilating hyaluronic acid. Unlike B. thetaiotaomicron, H. hathewayi N2-326 could not utilize heparin. The chondroitin lyase activity of H. hathewayi N2-326 was found to be present predominantly in the culture supernatant. Genome sequence analysis revealed three putative GAG lyases, but only the HH-chondroitin ABC lyase was upregulated in the presence of CSA. In addition, five CAZyme gene clusters containing GAG metabolism genes were significantly upregulated when grown on CSA. Further characterization of the recombinant HH-chondroitin ABC lyase revealed that it cleaves GAGs predominantly in an exo-mode to produce unsaturated disaccharides as the primary hydrolytic product while exhibiting a higher specific activity than reported chondroitin ABC lyases. HH-chondroitin ABC lyase represents the first characterized chondroitin lyase from intestinal Firmicutes and offers a viable commercial option for the production of chondroitin, dermatan, and hyaluronan oligosaccharides and also for potential medical applications. IMPORTANCE An increased understanding of GAG metabolism by intestinal bacteria is critical in identifying the driving factors for the composition, modulation, and homeostasis of the human gut microbiota. In addition, GAG-depolymerizing polysaccharide lyases are highly desired enzymes for the production of GAG oligosaccharides and as therapeutics. At present, the dissection of the enzymatic machinery for GAG degradation is highly skewed toward Bacteroides. In this study, we have isolated an efficient GAG-degrading Firmicutes bacterium from human feces and characterized the first chondroitin ABC lyase from a Firmicutes, which complements the fundamental knowledge of GAG utilization in the human colon. The genomic and transcriptomic analysis of the bacterium shows that Firmicutes might use a distinct approach to catabolize GAGs from that used by Bacteroides. The high specific activity of the characterized chondroitin ABC lyase aids future attempts to develop a commercial chondroitinase for industrial and medicinal applications.


Subject(s)
Chondroitin ABC Lyase , Glycosaminoglycans , Humans , Bacteroides/genetics , Bacteroides/metabolism , Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Firmicutes/metabolism , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Oligosaccharides/chemistry , Substrate Specificity , Intestines/metabolism
3.
Enzyme Microb Technol ; 143: 109701, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33375969

ABSTRACT

Chondroitinase ABC can be used to prepare chondroitin sulfate (CS) oligosaccharides efficiently and environmentally. It also promotes nerve recovery through enzymatic degradation of glycosaminoglycan chains in damaged nerve tissue. In this study, two new chondroitin sulfate ABC lyases were expressed and characterized from Edwardsiella tarda LMG2793, with molecular weight of 116.8 kDa and 115.9 kDa, respectively. Two lyases ChABC I and ChABC II belonged to the polysaccharide lyase (PL) family 8. ChABC I and ChABC II showed enzyme activity towards chondroitin sulfate A (CS-A), CS-B, CS-C and CS-D, but had no activity towards hyaluronan (HA). The optimal temperature for ChABC I to exhibit the highest activity against CS-A was 40 °C and the optimal pH was 7.0. ChABC II showed the highest activity to CS-A at optimal temperature of 40 °C and pH of 9.0. ChABC I and ChABC II were stable at 37 °C and remained about 90 % of activity after incubation at 37 °C for 3 h. Many metal ions had no effect on the activity of ChABC I and ChABC II. These properties were beneficial to their further basic research and application. ChABC I was an endo-type enzyme while ChABC II was an exo-type enzyme. A group of amino acids were selected for further study by evaluating the sequence homology with other CS degradation lyases. Mutagenesis studies speculated that the catalytic residues in ChABC I were His522, Tyr529 and Arg581. The catalytic residues of ChABC II were His498, Tyr505 and Arg558. This work will contribute to the structural and functional characterization of biomedically relevant CS and promote the application of CS lyase in further basic research and therapeutics.


Subject(s)
Chondroitin ABC Lyase , Chondroitin Sulfates , Chondroitin ABC Lyase/genetics , Cloning, Molecular , Edwardsiella tarda/genetics , Ions
4.
Biotechnol J ; 16(5): e2000321, 2021 May.
Article in English | MEDLINE | ID: mdl-33350041

ABSTRACT

Chondroitinase ABC I (csABC I) has attracted intensive attention because of its great potential in heparin refining and the enzymatic preparation of low-molecular-weight chondroitin sulfate (LMW-CS). However, low thermal resistance (<30℃) restricts its applications. Herein, structure-guided and sequence-assisted combinatorial engineering approaches were applied to improve the thermal resistance of Proteus vulgaris csABC I. By integrating the deletion of the flexible fragment R166-L170 at the N-terminal domain and the mutation of E694P at the C-terminal domain, variant NΔ5/E694P exhibited 247-fold improvement of its half-life at 37℃ and a 2.3-fold increase in the specific activity. Through batch fermentation in a 3-L fermenter, the expression of variant NΔ5/E694P in an Escherichia coli host reached 1.7 g L-1 with the activity of 1.0 × 105 U L-1 . Finally, the enzymatic approach for the preparation of LMW-CS was established. By modulating enzyme concentration and controlling depolymerization time, specifically distributed LMW-CS (7000, 3400, and 1900 Da) with low polydispersity was produced, demonstrating the applicability of these processes for the industrial production of LMW-CS in a more environmentally friendly way.


Subject(s)
Chondroitin ABC Lyase , Chondroitin Sulfates , Chondroitin ABC Lyase/genetics , Chondroitinases and Chondroitin Lyases , Molecular Weight , Proteus vulgaris/genetics
5.
Int J Biol Macromol ; 164: 3762-3770, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32871123

ABSTRACT

Chondroitinases degrade chondroitin sulfate (CS) into oligosaccharides, of which the biological activities have vital roles in various fields. Some chondroitinases in polysaccharide lyase family 8 (PL8) have been classified into four subfamilies (PL8_1, PL8_2, PL8_3, and PL8_4) based on their sequence similarity and substrate specificities. In this study, a gene, vpa_0049, was cloned from marine bacterium Vibrio sp. QY108. The encoded protein, Vpa_0049, did not belong to the four existing subfamilies in PL8 based on phylogenetic analysis. Vpa_0049 could degrade various glycosaminoglycans (CS-A, CS-B, CS-C, CS-D, and HA) into unsaturated disaccharides in an endolytic manner, which was different from PL8 lyases of four existing subfamilies. The maximum activity of Vpa_0049 on different glycosaminoglycan substrates appeared at 30-37 °C and pH 7.0-8.0 in the presence of NaCl. Vpa_0049 showed approximately 50% of maximum activity towards CS-B and HA at 0 °C. It was stable in alkaline conditions (pH 8.0-10.6) and 0-30 °C. Our study provides a new broad-substrate chondroitinase and presents an in-depth understanding of PL8.


Subject(s)
Chondroitin ABC Lyase/genetics , Cloning, Molecular , Polysaccharide-Lyases/genetics , Vibrio/genetics , Chondroitin Lyases/genetics , Chondroitin Sulfates/genetics , Glycosaminoglycans/genetics , Oligosaccharides/genetics , Phylogeny , Substrate Specificity , Vibrio/enzymology
6.
Biochem Biophys Res Commun ; 532(3): 420-426, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32888649

ABSTRACT

The efficiency of cell therapy after spinal cord injury (SCI) depend on the survival of transplanted cells. However, sterile microenvironment and glial scar hyperplasia extremely reduce their numbers. Our previous study found overexpression of ChABC gene is positively correlated to migration ability. Expression of PTEN gene is closely associated with proliferation. However, whether manipulation of PTEN and ChABC on adipose-derived mesenchymal stem cells (ADSCs) promote motor recovery is unknown. This study aimed to promote hindlimb function recovery in SCI rats by enhancing proliferation and migration ability of ADSCs, transiently silencing expression of PTEN following overexpression of ChABC (double-gene modified ADSCs, DG-ADSCs). After PTEN silencing, we observed strong proliferation and accelerated G1-S transition in DG-ADSCs using CCK8 assay and flow cytometry. In addition, we demonstrated that migration numbers of DG-ADSCs were higher than control group using Transwell assay. The protein and mRNA levels of MAP2 and ßⅢ-tubulin in DG-ADSCs were increased compared with ADSCs. These results were further confirmed in SCI rats. Increased survival cells and reduction of glial scars were quantitatively analyzed in DG-ADSCs groups, which is definitely correlated to function recovery. Recovery of motor function was observed in DG-ADSCs treatment rats using BBB score, which emphasized that improved viability of transplanted cells and reduction of glial scars were an effective strategy for enhancing recovery of neurological function after SCI.


Subject(s)
Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/metabolism , Mesenchymal Stem Cell Transplantation , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Spinal Cord Injuries/therapy , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Gene Silencing , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/metabolism , Neurons/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Up-Regulation
7.
FASEB J ; 34(8): 10605-10622, 2020 08.
Article in English | MEDLINE | ID: mdl-32543730

ABSTRACT

Ventral root avulsion leads to severe motoneuron degeneration and prolonged distal nerve denervation. After a critical period, a state of chronic denervation develops as repair Schwann cells lose their pro-regenerative properties and inhibitory factors such as CSPGs accumulate in the denervated nerve. In rats with ventral root avulsion injuries, we combined timed GDNF gene therapy delivered to the proximal nerve roots with the digestion of inhibitory CSPGs in the distal denervated nerve using sustained lentiviral-mediated chondroitinase ABC (ChABC) enzyme expression. Following reimplantation of lumbar ventral roots, timed GDNF-gene therapy enhanced motoneuron survival up to 45 weeks and improved axonal outgrowth, electrophysiological recovery, and muscle reinnervation. Despite a timed GDNF expression period, a subset of animals displayed axonal coils. Lentiviral delivery of ChABC enabled digestion of inhibitory CSPGs for up to 45 weeks in the chronically denervated nerve. ChABC gene therapy alone did not enhance motoneuron survival, but led to improved muscle reinnervation and modest electrophysiological recovery during later stages of the regeneration process. Combining GDNF treatment with digestion of inhibitory CSPGs did not have a significant synergistic effect. This study suggests a delicate balance exists between treatment duration and concentration in order to achieve therapeutic effects.


Subject(s)
Chondroitin ABC Lyase/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Nerve Regeneration/genetics , Spinal Nerve Roots/physiology , Animals , Axons/physiology , Cell Line , Female , Genetic Therapy/methods , HEK293 Cells , Humans , Motor Neurons/physiology , Nerve Regeneration/physiology , Rats , Rats, Wistar , Recovery of Function/genetics , Schwann Cells/physiology
8.
Int J Nanomedicine ; 15: 315-332, 2020.
Article in English | MEDLINE | ID: mdl-32021182

ABSTRACT

PURPOSE: The clinical outcome of spinal cord injury is usually poor due to the lack of axonal regeneration and glia scar formation. As one of the most classical supporting cells in neural regeneration, Schwann cells (SCs) provide bioactive substrates for axonal migration and release molecules that regulate axonal growth. However, the effect of SC transplantation is limited by their poor migration capacity in the astrocyte-rich central nervous system. METHODS: In this study, we first magnetofected SCs with chondroitinase ABC-polyethylenimine functionalized superparamagnetic iron oxide nanoparticles (ChABC/PEI-SPIONs) to induce overexpression of ChABC for the removal of chondroitin sulfate proteoglycans. These are inhibitory factors and forming a dense scar that acts as a barrier to the regenerating axons. In vitro, we observed the migration of SCs in the region of astrocytes after the application of a stable external magnetic field. RESULTS: We found that magnetofection with ChABC/PEI-SPIONs significantly up-regulated the expression of ChABC in SCs. Under the driven effect of the directional magnetic field (MF), the migration of magnetofected SCs was enhanced in the direction of the magnetic force. The number of SCs with ChABC/PEI-SPIONs migrated and the distance of migration into the astrocyte region was significantly increased. The number of SCs with ChABC/PEI-SPIONs that migrated into the astrocyte region was 11.6- and 4.6-fold higher than those observed for the intact control and non-MF groups, respectively. Furthermore, it was found that SCs with ChABC/PEI-SPIONs were in close contact with astrocytes and no longer formed boundaries in the presence of MF. CONCLUSION: The mobility of the SCs with ChABC/PEI-SPIONs was enhanced along the axis of MF, holding the potential to promote nerve regeneration by providing a bioactive microenvironment and relieving glial obstruction to axonal regeneration in the treatment of spinal cord injury.


Subject(s)
Astrocytes/physiology , Chondroitin ABC Lyase/metabolism , Magnetite Nanoparticles/therapeutic use , Nerve Regeneration/physiology , Schwann Cells/physiology , Animals , Astrocytes/cytology , Axons/drug effects , Cell Movement , Cells, Cultured , Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/metabolism , Female , Magnetic Fields , Magnetite Nanoparticles/chemistry , Male , Nerve Regeneration/drug effects , Polyethyleneimine/chemistry , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Spinal Cord Injuries/therapy
9.
PLoS One ; 15(1): e0221851, 2020.
Article in English | MEDLINE | ID: mdl-31961897

ABSTRACT

BACKGROUND: There is currently no effective treatment for promoting regeneration of injured nerves in patients who have sustained injury to the central nervous system such as spinal cord injury. Chondroitinase ABC is an enzyme, which promotes neurite outgrowth and regeneration. It has shown considerable promise as a therapy for these conditions. The aim of the study is to determine if targeting chondroitinase ABC expression to the neuronal axon can further enhance its ability to promote axon outgrowth. Long-distance axon regeneration has not yet been achieved, and would be a significant step in attaining functional recovery following spinal cord injury. METHODOLOGY/PRINCIPAL FINDINGS: To investigate this, neuronal cultures were transfected with constructs encoding axon-targeted chondroitinase, non-targeted chondroitinase or GFP, and the effects on neuron outgrowth and sprouting determined on substrates either permissive or inhibitory to neuron regeneration. The mechanisms underlying the observed effects were also explored. Targeting chondroitinase to the neuronal axon markedly enhances its ability to promote neurite outgrowth. The increase in neurite length is associated with an upregulation of ß-integrin staining at the axonal cell surface. Staining for phosphofocal adhesion kinase, is also increased, indicating that the ß-integrins are in an activated state. Expression of chondroitinase within the neurons also resulted in a decrease in expression of PTEN and RhoA, molecules which present a block to neurite outgrowth, thus identifying two of the pathways by which ChABC promotes neurite outgrowth. CONCLUSIONS / SIGNIFICANCE: The novel finding that targeting ChABC to the axon significantly enhances its ability to promote neurite extension, suggests that this may be an effective way of promoting long-distance axon regeneration following spinal cord injury. It could also potentially improve its efficacy in the treatment of other pathologies, where it has been shown to promote recovery, such as myocardial infarction, stroke and Parkinson's disease.


Subject(s)
Chondroitin ABC Lyase/genetics , Nerve Regeneration/genetics , Neuronal Outgrowth/genetics , Spinal Cord Injuries/genetics , Animals , Axons/metabolism , Chondroitin ABC Lyase/antagonists & inhibitors , Gene Expression Regulation/genetics , Humans , Neurites/metabolism , Neurons/metabolism , Neurons/physiology , PTEN Phosphohydrolase/genetics , Recovery of Function/genetics , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , rhoA GTP-Binding Protein/genetics
10.
J Biotechnol ; 309: 131-141, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-31935418

ABSTRACT

Chondroitinase ABC I (cABC I) has received notable attention in treatment of spinal cord injuries and its application as therapeutics has been limited due to low thermal stability at physiological temperature. In this study, cABC I enzyme was immobilized on the dextran-coated Fe3O4 nanoparticles through physical adsorption to improve the thermal stability. The nanoparticles were characterized using XRD, SEM, VSM, and FTIR analyses. Response surface methodology and central composite design were employed to assess factors affecting the activity of immobilized cABC I. Experimental results showed that pH 6.3, temperature 24 °C, enzyme/support mass ratio 1.27, and incubation time 5.7 h were the optimal immobilization conditions. It was found that thermal stability of immobilized cABC I was significantly improved. In-vitro cABC I release was studied under pH 7.5 and temperature 37 °C and the results indicated that 70 % release occurred after 9 h and the release mechanism was first-order kinetic model.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Dextrans/chemistry , Enzymes, Immobilized/chemistry , Magnetite Nanoparticles/chemistry , Adsorption , Chondroitin ABC Lyase/genetics , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Kinetics , Proteus vulgaris/genetics , Temperature , X-Ray Diffraction
11.
Int J Biol Macromol ; 143: 41-48, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31786296

ABSTRACT

Chondroitinase ABC I (ChSase ABC I) is a key enzyme of chondroitin sulfate (CS) degradation and widely used for CS detection in the medicine filed. However, the recombinant ChSase ABC I was weakly expressed in Escherichia coli because the forms of it were mostly inclusion bodies. In this study, a signal peptide (pelB) was used for the soluble form expression of ChSase ABC I in E. coli. Then the culture condition for ChSase ABC I expression was optimized through response surface methodology. Results revealed that the expression level of ChSase ABC I in a 7.5 L fermentor (29.03 mL-1) was approximately 1.65-fold higher than that of the shake flask level (17.55 mL-1). The enzymatic properties and kinetic constants of recombinant ChSase ABC I were also studied. Recombinant ChSase ABC I was also used to detect the specific disaccharides content of CS from different sources. This study not only eliminates the problem of the enzyme expressed as an inclusion body, but also solves the current problem of expensive ChSase ABC. In a word, it would be an ideal strategy for ChSase ABC high-efficiency expression and a great method to detect specific disaccharides of CS in biomedical field.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/genetics , Chondroitin Sulfates/analysis , Disaccharides/analysis , Chemical Phenomena , Chondroitin ABC Lyase/isolation & purification , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfates/chemistry , Chromatography, High Pressure Liquid , Disaccharides/chemistry , Fermentation , Hydrogen-Ion Concentration , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Temperature
12.
Carbohydr Polym ; 224: 115135, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31472845

ABSTRACT

Chondroitin sulfate ABC lyases (csABCs) have attracted intensive attention because of their wide potential applications in promoting tissue regeneration and generating oligosaccharides. In the present study, three csABC I encoding sequences were analyzed and site-directed mutagenesis results demonstrate that residues Leu125 and Leu322 are essential to activity and mutation of each leucine residue to proline dramatically decreased enzymatic activity. Additionally, our results showed that mutation of I309 V significantly increased the catalytic efficiency. By recruiting OmpA signal peptide and engineering the permeability of cell membrane with deletion of a lipoprotein encoding gene lpp, all recombinant enzymes were secreted and the extracellular activity was finally increased to 2.99 ±â€¯0.1 U/mL in batch fermentation. More importantly, the engineered csABC I with high activity can rapidly degrade chondroitin sulfate to the end tetrasaccharides and disaccharides, demonstrating its applicability for preparation of chondroitin sulfate oligosaccharides.


Subject(s)
Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfates/chemistry , Oligosaccharides/chemistry , Protein Engineering , Amino Acid Sequence , Biocatalysis , Chondroitin ABC Lyase/chemistry , Gene Expression , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Proteus vulgaris/enzymology
13.
Arch Biochem Biophys ; 668: 46-53, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31103558

ABSTRACT

Chondroitinase ABC I (cABC I) can degrade inhibitory molecules for axon regrowth at the site of damage after spinal cord injury (SCI). One of the main problems in the practical application is the possibility of structural changes that lead to the inactivation of the enzyme. In current work, three variants of cABC I was designed and constructed by manipulation of a short helix conformation (Gln678-Leu679-Ser680-Gln681); where Gln residues were converted to Glu. According to the enzyme kinetics studies, the catalytic efficiency of the Q681E and double mutant (Q678E/Q681E) increases in comparison with WT enzyme; while that of Q678E decreases. It was also shown that the rate of the inactivation of the enzyme variants over time is greater in WT and Q678E variants than that of the Q681E and double mutant. Negative values of entropy change of thermal inactivation measurements; demonstrate that inactivation of the WT and Q678E variants are mainly originated from aggregation. These observations can be explained by considering the repulsive electrostatic interaction between enzyme molecules that prevents protein aggregation over time. It is concluded that increasing the solubility of the Q681E and double mutant via favorable interactions of surface-exposed charged residues with dipole momentum of water molecules accompanied by the presence of intermolecular repulsive electrostatic interaction leads to decreasing the rate of aggregation in both long-term storage and heat-induced structural changes.


Subject(s)
Bacterial Proteins/metabolism , Chondroitin ABC Lyase/metabolism , Protein Aggregates/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/genetics , Enzyme Stability , Escherichia coli/genetics , Glutamic Acid/chemistry , Glutamine/chemistry , Kinetics , Mutagenesis, Site-Directed , Protein Conformation , Protein Domains/genetics , Protein Multimerization/genetics , Proteus vulgaris/enzymology , Thermodynamics
14.
Protein J ; 38(2): 151-159, 2019 04.
Article in English | MEDLINE | ID: mdl-30859376

ABSTRACT

Immune response stimulation and inactivation of chondroitinase ABC I in physiological condition have been limited its use in various clinical conditions as a bacterial enzyme drug. In the present study, we have investigated some structural and functional features of N∆89, C∆274 and N∆89C∆274; three designed truncated cABC I, in order to clarify the unclear role of two terminal parts of cABC I i.e., the 1-89 and 747-1021 amino acids sequences of the full length enzyme through truncation. As a result, the numbers of potential epitopes, the susceptibility to trypsin digestion, ANS fluorescence spectra, and fluorescence quenching using KI and acrylamide were diminished for N∆89 and C∆274 in comparison to the wild type. Secondary and tertiary structure investigation for N∆89 and C∆274 revealed that the intrinsic fluorescence was increased and Far-UV CD spectra were changed accordingly. Relative to the wild type enzyme, 0.164, 0.195 remaining activity and lack of activity was shown with the zymographic assay for N∆89, C∆274 and N∆89C∆274 variants, respectively. The diminished enzyme activity and structural changes suggested a reorientation of microenvironments interactions including cation-π interactions around structural elements toward lowering regional mobility. Constructing applicable truncated cABC I with improved features could be regarded as a strategy to regain new possible functional advantages over the full length enzyme.


Subject(s)
Bacterial Proteins/chemistry , Chondroitin ABC Lyase/chemistry , Bacterial Proteins/genetics , Chondroitin ABC Lyase/genetics , Enzyme Stability , Escherichia coli/genetics , Kinetics , Models, Molecular , Mutation , Protein Conformation , Proteus vulgaris/enzymology
15.
J Control Release ; 297: 14-25, 2019 03 10.
Article in English | MEDLINE | ID: mdl-30690102

ABSTRACT

Central nervous system (CNS) injuries, such as stroke and spinal cord injuries, result in the formation of a proteoglycan-rich glial scar, which acts as a barrier to axonal regrowth and limits the regenerative capacity of the CNS. Chondroitinase ABC (ChABC) is a potent bacterial enzyme that degrades the chondroitin sulfate proteoglycan (CSPG) component of the glial scar and promotes tissue recovery; however, its use is significantly limited by its inherent instability at physiological temperatures. Here, we demonstrate that ChABC can be stabilized using site-directed mutagenesis and covalent modification with poly(ethylene glycol) chains (i.e. PEGylation). Rosetta protein structure modeling was used to screen >20,000 single point mutations, and four potentially stabilizing mutations were tested in vitro. One of the mutations, N1000G (asparagine ➔ glycine at residue 1000), significantly improved the long-term activity of the protein, doubling its functional half-life. PEGylation of this ChABC mutant inhibited unfolding and aggregation and resulted in prolonged bioactivity with a 10-fold increase in activity compared to the unmodified protein after two days. Local, affinity-controlled release of the modified protein (PEG-N1000G-ChABC) was achieved by expressing it as a fusion protein with Src homology 3 (SH3) and delivering the protein from a methylcellulose hydrogel modified with SH3 binding peptides. This affinity-based release strategy provided sustained PEG-N1000G-ChABC-SH3 release over several days in vitro. Direct implantation of the hydrogel delivery vehicle containing stabilized PEG-N1000G-ChABC-SH3 onto the rat brain cortex in a sub-acute model of stroke resulted in significantly reduced CSPG levels in the penumbra of 49% at 14 and 40% at 28 days post-injury compared to animals treated with the vehicle alone.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Nanocapsules/chemistry , Nerve Regeneration/drug effects , Polyethylene Glycols/chemistry , Stroke/metabolism , Animals , Axons/metabolism , Brain/drug effects , Chondroitin ABC Lyase/genetics , Chondroitin Sulfate Proteoglycans/drug effects , Drug Liberation , Male , Mutagenesis/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuroglia/metabolism , Proteus vulgaris/enzymology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , src Homology Domains
16.
Int J Biol Macromol ; 119: 779-784, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30081121

ABSTRACT

Chondroitinase ABC I (ChSase ABC I), as a polysaccharide lyase, can catalyze high molecular weight chondroitin sulfate (CS) to low molecular weight glycosaminoglycan which are easier to be absorbed and utilized by organisms. In this study, to enhance the production of ChSase ABC I and avoid the negative influence of tags on its catalytic efficiency, we employed molecular chaperones to co-express with ChSase ABC I. Firstly, different molecular chaperones and their combinations were screened and GroES exhibited the best positive effect. Consecutively, fermentation conditions were optimized to further improve the production. As a result, the production of ChSase ABC I was increased to 4640.44 ±â€¯896.26 IU/g wet weight, a 2.15-fold higher value when compared with that of control in the same fermentation conditions. After that, to testify the influence of GroES on characterization of ChSase ABC I, the optimal pH and temperature, and kinetic parameters were confirmed. The affinity to substrate of ChSase ABC I with GroES assist was increased 7 folds as compared to the native ChSase ABC I, and ChSase ABC I with GroES co-expression still has high catalytic activity. This work not only presents to date the first achievement of ChSase ABC I high-level production with molecular chaperone co-expression, but also serves as a potential basis for its industrial application.


Subject(s)
Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Molecular Chaperones/genetics , Fermentation , Hydrogen-Ion Concentration , Kinetics , Protein Folding , Temperature
17.
Int J Biol Macromol ; 116: 811-816, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29777817

ABSTRACT

Previously, we attempted to improve the thermostability of chondroitinase ABC I by substituting proline in flexible sites and successfully obtained a mutant; E138P, with increased thermostability (Kheirollahi et al., 2017). In this study, we focused on the role of Glu138 in activity and stability of the enzyme using its further mutation to Ala, Lys, and Asp. Moreover, we coupled the two mutations E138P and Q140A, whose stabilizing effects were reported previously, and evaluated their simultaneous effects on activity, stability, and structure of the enzyme. The results indicate that substitution of Glu138 with the above-mentioned amino acids changed kinetic properties of cABC I but did not lead to increased stability. Moreover, replacement of Glu138 with Lys and Asp caused significant structural changes. These findings lead to the tentative conclusion that improvement in thermal stability of E138P variant is due to the stabilizing effect of proline at position 138. In addition, the double variant showed a significant increase in catalytic efficiency, howbeit its kinetic stability decreased. Moreover, structural analysis of the double mutant form revealed that its tertiary and secondary structure content decreased partially, while its structural flexibility increased.


Subject(s)
Amino Acid Substitution , Chondroitin ABC Lyase , Hot Temperature , Mutation, Missense , Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/genetics , Enzyme Stability/genetics , Protein Structure, Secondary
18.
Appl Biochem Biotechnol ; 186(2): 358-370, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29627885

ABSTRACT

Removal of chondroitin sulfate glycosaminoglycan (GAG) chains with chondroitinase ABC I (chABC I) in CNS injury models promotes both saxon regeneration and plasticity. It has been suggested that direct interaction between an aromatic pair appears to contribute about - 1.3 kcal/mol to the stability of a folded protein, so introducing an aromatic pair by point mutation might increase the enzyme activity and thermal stability as in the case of mesophilic xylanase, although using this approach destabilized T4 lysozyme. In this study, we used site-directed mutagenesis to investigate the effect of new aromatic pairs on activity and stability of chABC I. We replaced Ile295, Ser581, and Gly730 adjacent to pre-existing aromatic residues with Tyr to obtain new aromatic pairs, i.e., Tyr295/His372, Tyr576/Tyr581, and Tyr623/Tyr730. Results showed that Km values of S581Y and G730Y variants decreased relative to wild-type enzyme while their catalytic efficiency (kcat/Km) increased but I295Y variant was inactive. Also, long-term and thermal stability of the active mutants was decreased. Fluorescence and circular dichroism studies showed that these mutations resulted in a more flexible enzyme structures: a finding which was confirmed by thermal and limited proteolytic studies. In conclusion, the activity of chABC I can be improved by introducing appropriate aromatic pairs at the enzyme surface. This approach did not provide any promising results regarding the enzyme stability.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Catalysis , Chondroitin ABC Lyase/genetics , Circular Dichroism , Enzyme Stability , Mutagenesis, Site-Directed , Protein Structure, Secondary , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Thermodynamics
19.
J Biomol Struct Dyn ; 36(3): 679-688, 2018 02.
Article in English | MEDLINE | ID: mdl-28278758

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are potent inhibitors of growth in the adult central nervous system. Use of the enzyme chondroitinase ABC I (ChABC I) as a strategy to reduce CSPG inhibition in experimental models of spinal cord injury has led to observations of its remarkable capacity for repair. More importantly, ChABC therapy has been demonstrated to promote significant recovery of function to spinal injured animals. Despite this incomparable function of ChABC I, its clinical application has been limited because of its thermal instability as reported in the literature. In a recent study by Nazari-Robati et al., thermal stability of ChABC I was improved by protein engineering using site-directed mutagenesis method. Here, in this study, molecular dynamics simulations were used to take a closer look into the phenomenon leading to the experimentally observed thermal stability improvement followed by the corresponding site-directed mutagenesis. We concluded that the mutations induce local flexibility along with a re-conformation into the native structure which consequently increase the protein thermal stability.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin Sulfate Proteoglycans/chemistry , Enzyme Stability , Spinal Cord Injuries/enzymology , Animals , Central Nervous System/drug effects , Chondroitin ABC Lyase/genetics , Chondroitin Sulfate Proteoglycans/biosynthesis , Disease Models, Animal , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Temperature
20.
Enzyme Microb Technol ; 107: 64-71, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28899488

ABSTRACT

Chondroitin Sulfate Proteoglycans (CSPGs) are the main inhibitors for axon regeneration after damaging of Central Nervous System (CNS). Chondroitinase ABC I (cABC I) can degrade CSPGs by removing chondroitin and dermatan sulfate side chains from proteoglycans. Hence, it may be considered as an attractive candidate in biomedicine. For practical applications of this enzyme, increasing the effective circulating level and reducing the number and volume of injections for patients is one of the main concerns which is directly related to conformational stability and catalytic efficiency of the enzyme. Structural examination of C-terminal domain of cABC I reveals that there are a few numbers of residues in helical conformation which are positioned at the context of a cohesive structural organization of ß-strands. In line with our previous studies on C-terminal domain of cABC I and regarding the residues in α-helix conformation; we designed and constructs some representative mutants including M889K, M889L, L679D/M889K and L679S/M889K. According to structural and functional characterization of protein variants and regarding the wide range of variability in determining parameters for ß-sheet conformation, we proposed a model in which the structural integrity of ß-strands at C-terminal domain can be manipulated and directed toward a new patterns of organization, some of them may have positive effects on the structural and functional features of the enzyme. Using this strategy it may be possible to improve functional and structural features of the enzyme by engineering the intra-molecular interactions in positions far from the active site of the enzyme.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biotechnology , Chondroitin ABC Lyase/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Enzyme Stability , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Domains , Protein Engineering , Proteus vulgaris/enzymology , Proteus vulgaris/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL